NLR Members in Inflammation-Associated

Total Page:16

File Type:pdf, Size:1020Kb

NLR Members in Inflammation-Associated Cellular & Molecular Immunology (2017) 14, 403–405 & 2017 CSI and USTC All rights reserved 2042-0226/17 $32.00 www.nature.com/cmi RESEARCH HIGHTLIGHT NLR members in inflammation-associated carcinogenesis Ha Zhu1,2 and Xuetao Cao1,2,3 Cellular & Molecular Immunology (2017) 14, 403–405; doi:10.1038/cmi.2017.14; published online 3 April 2017 hronic inflammation is regarded as an impor- nucleotide-binding and oligomerization domain IL-2,8 and NAIP was found to regulate the STAT3 Ctant factor in cancer progression. In addition (NOD)-like receptors (NLRs). TLRs and CLRs are pathway independent of inflammasome formation.9 to the immune surveillance function in the early located in the plasma membranes, whereas RLRs, The AOM/DSS model is the most popular model stage of tumorigenesis, inflammation is also known ALRs and NLRs are intracellular PRRs.3 Unlike used to study the function of NLRs in fl fl as one of the hallmarks of cancer and can supply other families that have been shown to bind their in ammation-associated carcinogenesis. In amma- the tumor microenvironment with bioactive mole- specific cognate ligands, the distinct ligands for somes initiated by NLRs or AIM2 have been widely cules and favor the development of other hallmarks NLRs are still unknown. In fact, mounting evidence reported to participate in the maintenance of 10,11 Nlrp3 Nlrp6 of cancer, such as genetic instability and angiogen- suggests that NLRs function as cytoplasmic sensors intestinal homeostasis. -/-, -/-, Nlrc4 Nlrp1 Nlrx1 Nlrp12 esis. Moreover, inflammation contributes to the and participate in modulating TLR, RLR and CLR -/-, -/-, -/- and -/- mice are 4 more susceptible to AOM/DSS-induced colorectal changing tumor microenvironment by altering signaling pathways. NLRs can sense stimulatory cancer than wild-type (WT) mice.11 Loss of IL-18 stromal cell turnover rates and polarizing immune signals from pathogens, host cells and environmen- may partially account for the enhanced tumorigen- cell immunosuppressive capabilities.1 In addition, tal sources, although the interaction patterns are 3 esis in the abovementioned mice. IL-18 production many tumor suppressor genes have been reported still unclear. Importantly, dysfunction of NLRs fl is mainly regulated by inflammasomes and can to be involved in inflammation modulation.2 How- leads to various in ammatory diseases, autoim- fl stimulate regeneration and repair in the colon.4 ever, the types of molecules and the underlying mune diseases and in ammation-associated fl carcinogenesis.5 Given the vital roles of NLRs in Molecules that can regulate the functions of the mechanisms involved in in ammation-associated fl inflammation and disease progression, research is in ammasome are currently gaining attention and cancer need to be further investigated, which may fi now focused on elucidating their roles in the are potential bene cial treatments for provide new targets for the control of cancer. inflammasome-associated disorders.12 Moreover, pathogenesis of diseases such as cancer, with the Recognition of danger-associated molecular pat- deficiency of NLRX1, NOD1 or NOD2, which are aim of finding new therapeutic targets. terns (DAMPs) and pathogen-associated molecular non-inflammasome NLRs, results in increased NLRs contain three conserved functional patterns (PAMPs) by pattern recognition receptors susceptibility to colorectal cancer. NLRX1 sensitizes fl domains: a C-terminal leucine-rich repeat for (PRRs) is the key step that modulates in amma- cells to apoptotic cell death in response to DSS ligand recognition, a central NACHT domain tion. Five major PRR families have been reported to treatment and promotes epithelial proliferation. essential for oligomerization and an N-terminal be involved in tumorigenesis, including the Toll- NLRX1 is a negative regulator of AKT and NF- protein-protein interaction domain. According to like receptors (TLRs), C-type lectin receptors κB. NOD1 participates in the maintenance of the the types of N-terminal domains, mammalian (CLRs), RIG-I-like receptors (RLRs), absent-in- intestinal epithelial barrier permeability and bal- NLRs are classified into NLRA, NLRB, NLRC and melanoma (AIM)-like receptors (ALRs) and the ances inflammatory responses under AOM/DSS NLRP subfamilies with acidic transactivation stimulation, whereas NOD2 regulates apoptosis domains, baculoviral inhibitory repeat (BIR)-like and the NF-κB and MAPK signaling pathways.11 1 Institute of Immunology, Zhejiang University domains, caspase recruitment domains and pyrin The functions of other non-inflammasome NLRs in 4,6 fi School of Medicine, Hangzhou 310058, China; domains. Since the rst NLR was discovered in inflammation-associated carcinogenesis are 2National Key Laboratory of Medical Immunology 7 early 2000, at least 23 distinct NLR and NLR-like unknown. Recently, Dr. Rajendra Karki’sresearch & Institute of Immunology, Second Military Med- fi 4 proteins have been identi ed in humans. NLRs, group published a paper in Nature that described ical University, Shanghai 200433, China and including NLRP1, NLRP3, NLRP6, NLPR7, 3 the function of a poorly researched non- Department of Immunology & Center for Immu- NLRP12, NLRC4 and NAIP, can function in an fl notherapy, Institute of Basic Medical Sciences, in ammasome-forming NLR, NLRC3, in colitis- inflammasome-dependent manner to produce 13 Peking Union Medical College, Chinese Academy associated carcinogenesis. interleukin-1β (IL-1β), IL-18 and caspase-1, thus NLRC3 was first identified in 2005 and found to of Medical Sciences, Beijing 100005, China fl Correspondence: Professor X Cao, National Key inducing in ammation and regulating cell pyrop- be highly expressed in T lymphocytes. NLRC3 has Laboratory of Medical Immunology and Institute tosis. By contrast, other NLRs such NLRP10, been reported to suppress the signaling path- of Immunology, Second Military Medical Univer- NOD1, NOD2, NLRC3, NLRC5, NLRX1 and ways involved in T cell activation by influencing sity, 800 Xiang Yin Road, Shanghai 200433, CIITA function independent of the inflammasome the transcription factors NF-κB, NFAT and China. and can regulate NF-κB and MAPK pathways or act AP1.14 When stimulated by virus infection, NLRC3 E-mail: [email protected] as transcriptional regulators.6 Interestingly, the negatively regulates the STING pathway in bone Received: 6 February 2017; Accepted: 6 February functional patterns of NLRs are not fixed. Recently, marrow-derived macrophages by directly associat- 2017 NLRP3 has been reported to regulate STAT5 and ing with STING and blocking STING traffic- NLR members in carcinogenesis H Zhu and X Cao 404 king to puncta, thus inhibiting interferon-β expression.15 Reports on the functions of NLRC3 in carcinogenesis are rare. Karki et al.13 demon- strated that NLRC3 expression is significantly reduced in colorectal cancer tissues in mice; then, the authors established Nlrc3-/- mice and treated them with AOM/DSS. As expected, the Nlrc3-/- mice were more susceptible to the induction of colorectal cancer and exhibited greater weight loss, more tumor nodules, larger tumor sizes and higher histological scores. These findings indicate that NLRC3 acts as a suppressor in the development of colorectal cancer. Establishment of an inflammatory state is crucial for colitis-associated carcinogenesis.16 To assess the inflammatory state of the Nlrc3-/- mice treated with AOM/DSS, Karki et al.13 examined the expression of inflammatory cytokines, inflammatory signaling pathways and immune cell infiltration in colon tissues at days 80 and 14, which was 3 days after the first round of DSS treatment. There were no significant differences at day 80 between the Nlrc3-/- mice and WT mice. At day 14, pro- inflammatory cytokines, including IL-1β,IL-6, tumor necrosis factor, granulocyte colony stimulat- ing factor, chemokine (C-X-C motif) ligand 1, monocyte chemoattractant protein 1 and macro- phage inflammatory protein 1α,weregreatly increased in the Nlrc3-/- mice, whereas IL-18 remained unchanged. The NF-κB and STAT3 signaling pathways were activated at day 14, whereas the ERK signal pathway was unaltered. Thenumberofinnateimmunecells,including macrophages, neutrophils and natural killer cells, increased, whereas the numbers of B cells and T cells remained unchanged, and the number of dendritic cells decreased. These data demonstrate the hyper-inflammatory state of the Nlrc3-/- mice when treated with AOM/DSS, which possibly accounts for the hypersensitivity to carcinogenesis. Next, to further determine whether bone marrow- derived hematopoietic cells or stromal cells play a dominant role in the carcinogenesis of the Nlrc3-/- mice, Karki et al.13 established bone marrow chimeras and found that radioresistant stromal cells were more important than hematopoietic cells Figure 1 Functions of NLRs in inflammation-associated carcinogenesis. Under sustained during carcinogenesis, because cancer progression stimulation from pathogens, host cells or environmental sources, NLRs sense the was faster in Nlrc3-/- mice that received bone intracellular changes caused by infiltrating extracellular molecules or activation of the marrow from WT mice than in WT mice that downstream pathways of other PRRs and work to preserve cytosolic sanctity. NLRs function received bone marrow from Nlrc3-/- mice. The by associating with other proteins and through many interacting adaptors; however, details results from conditional knockout mice further are still unclear. NLRs can form inflammasomes to release IL-18 and IL-1β, which are pro- indicated that Nlrc3-/-intestinalepithelialcellswere
Recommended publications
  • Role of Nlrs in the Regulation of Type I Interferon Signaling, Host Defense and Tolerance to Inflammation
    International Journal of Molecular Sciences Review Role of NLRs in the Regulation of Type I Interferon Signaling, Host Defense and Tolerance to Inflammation Ioannis Kienes 1, Tanja Weidl 1, Nora Mirza 1, Mathias Chamaillard 2 and Thomas A. Kufer 1,* 1 Department of Immunology, Institute for Nutritional Medicine, University of Hohenheim, 70599 Stuttgart, Germany; [email protected] (I.K.); [email protected] (T.W.); [email protected] (N.M.) 2 University of Lille, Inserm, U1003, F-59000 Lille, France; [email protected] * Correspondence: [email protected] Abstract: Type I interferon signaling contributes to the development of innate and adaptive immune responses to either viruses, fungi, or bacteria. However, amplitude and timing of the interferon response is of utmost importance for preventing an underwhelming outcome, or tissue damage. While several pathogens evolved strategies for disturbing the quality of interferon signaling, there is growing evidence that this pathway can be regulated by several members of the Nod-like receptor (NLR) family, although the precise mechanism for most of these remains elusive. NLRs consist of a family of about 20 proteins in mammals, which are capable of sensing microbial products as well as endogenous signals related to tissue injury. Here we provide an overview of our current understanding of the function of those NLRs in type I interferon responses with a focus on viral infections. We discuss how NLR-mediated type I interferon regulation can influence the development of auto-immunity and the immune response to infection. Citation: Kienes, I.; Weidl, T.; Mirza, Keywords: NOD-like receptors; Interferons; innate immunity; immune regulation; type I interferon; N.; Chamaillard, M.; Kufer, T.A.
    [Show full text]
  • Inflammasome Activation-Induced Hypercoagulopathy
    cells Review Inflammasome Activation-Induced Hypercoagulopathy: Impact on Cardiovascular Dysfunction Triggered in COVID-19 Patients Lealem Gedefaw, Sami Ullah, Polly H. M. Leung , Yin Cai, Shea-Ping Yip * and Chien-Ling Huang * Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China; [email protected] (L.G.); [email protected] (S.U.); [email protected] (P.H.M.L.); [email protected] (Y.C.) * Correspondence: [email protected] (S.-P.Y.); [email protected] (C.-L.H.) Abstract: Coronavirus disease 2019 (COVID-19) is the most devastating infectious disease in the 21st century with more than 2 million lives lost in less than a year. The activation of inflammasome in the host infected by SARS-CoV-2 is highly related to cytokine storm and hypercoagulopathy, which significantly contribute to the poor prognosis of COVID-19 patients. Even though many studies have shown the host defense mechanism induced by inflammasome against various viral infections, mechanistic interactions leading to downstream cellular responses and pathogenesis in COVID-19 remain unclear. The SARS-CoV-2 infection has been associated with numerous cardiovascular disor- ders including acute myocardial injury, myocarditis, arrhythmias, and venous thromboembolism. The inflammatory response triggered by the activation of NLRP3 inflammasome under certain car- diovascular conditions resulted in hyperinflammation or the modulation of angiotensin-converting enzyme 2 signaling pathways. Perturbations of several target cells and tissues have been described in inflammasome activation, including pneumocytes, macrophages, endothelial cells, and dendritic cells. Citation: Gedefaw, L.; Ullah, S.; Leung, P.H.M.; Cai, Y.; Yip, S.-P.; The interplay between inflammasome activation and hypercoagulopathy in COVID-19 patients is an Huang, C.-L.
    [Show full text]
  • NLRP6 Induces Pyroptosis by Activation of Caspase-1 in Gingival
    JDRXXX10.1177/0022034518775036Journal of Dental ResearchNLRP6 Induces Pyroptosis 775036research-article2018 Research Reports: Biological Journal of Dental Research 2018, Vol. 97(12) 1391 –1398 © International & American Associations NLRP6 Induces Pyroptosis by Activation for Dental Research 2018 Article reuse guidelines: of Caspase-1 in Gingival Fibroblasts sagepub.com/journals-permissions DOI:https://doi.org/10.1177/0022034518775036 10.1177/0022034518775036 journals.sagepub.com/home/jdr W. Liu1* , J. Liu1*, W. Wang1, Y. Wang2,3, and X. Ouyang1 Abstract NLRP6, a member of the nucleotide-binding domain, leucine-rich repeat-containing (NLR) innate immune receptor family, has been reported to participate in inflammasome formation. Activation of inflammasome triggers a caspase-1–dependent programming cell death called pyroptosis. However, whether NLRP6 induces pyroptosis has not been investigated. In this study, we showed that NLRP6 overexpression activated caspase-1 and gasdermin-D and then induced pyroptosis of human gingival fibroblasts, resulting in release of proinflammatory mediators interleukin (IL)–1β and IL-18. Moreover, NLRP6 was highly expressed in gingival tissue of periodontitis compared with healthy controls. Porphyromonas gingivalis, which is a commensal bacterium and has periodontopathic potential, induced pyroptosis of gingival fibroblasts by activation of NLRP6. Together, we, for the first time, identified that NLRP6 could induce pyroptosis of gingival fibroblasts by activation of caspase-1 and may play a role in periodontitis. Keywords: periodontitis, pattern recognition receptors, cell death, Porphyromonas gingivalis, inflammasomes, flow cytometry Introduction have been demonstrated to participate in periodontitis (Huang et al. 2015; Chaves de Souza et al. 2016; Marchesan et al. Pyroptosis is a newly identified type of programmed cell death 2016).
    [Show full text]
  • Scholarly Commons NLRX1 Modulates Differentially NLRP3
    University of the Pacific Scholarly Commons Dugoni School of Dentistry Faculty Articles Arthur A. Dugoni School of Dentistry 10-1-2018 NLRX1 modulates differentially NLRP3 inflammasome activation and NF-κB signaling during Fusobacterium nucleatum infection Shu Chen Hung University of the Pacific Arthur A. Dugoni School of Dentistry Pei Rong Huang Chang Gung University Cassio Luiz Coutinho Almeida-Da-Silva University of the Pacific Arthur A. Dugoni School of Dentistry, [email protected] Kalina R. Atanasova University of Florida Ozlem Yilmaz Medical University of South Carolina See next page for additional authors Follow this and additional works at: https://scholarlycommons.pacific.edu/dugoni-facarticles Part of the Dentistry Commons Recommended Citation Hung, S., Huang, P., Almeida-Da-Silva, C. L., Atanasova, K. R., Yilmaz, O., & Ojcius, D. M. (2018). NLRX1 modulates differentially NLRP3 inflammasome activation and NF-κB signaling during Fusobacterium nucleatum infection. Microbes and Infection, 20(9-10), 615–625. DOI: 10.1016/j.micinf.2017.09.014 https://scholarlycommons.pacific.edu/dugoni-facarticles/705 This Article is brought to you for free and open access by the Arthur A. Dugoni School of Dentistry at Scholarly Commons. It has been accepted for inclusion in Dugoni School of Dentistry Faculty Articles by an authorized administrator of Scholarly Commons. For more information, please contact [email protected]. Authors Shu Chen Hung, Pei Rong Huang, Cassio Luiz Coutinho Almeida-Da-Silva, Kalina R. Atanasova, Ozlem Yilmaz, and David M. Ojcius This article is available at Scholarly Commons: https://scholarlycommons.pacific.edu/dugoni-facarticles/705 Version of Record: https://www.sciencedirect.com/science/article/pii/S1286457917301582 Manuscript_dd7f93413c97aff4865d54242a8b21e7 1 NLRX1 modulates differentially NLRP3 inflammasome activation 2 and NF-κB signaling during Fusobacterium nucleatum infection 3 4 5 Shu-Chen Hung 1, *, Pei-Rong Huang 2, Cássio Luiz Coutinho Almeida-da-Silva 1,3 , 6 Kalina R.
    [Show full text]
  • The Intestinal Parasite Cryptosporidium Is Controlled by an Enterocyte Intrinsic Inflammasome That Depends on NLRP6
    The intestinal parasite Cryptosporidium is controlled by an enterocyte intrinsic inflammasome that depends on NLRP6 Adam Saterialea,1, Jodi A. Gullicksruda, Julie B. Engilesa, Briana I. McLeoda, Emily M. Kuglera, Jorge Henao-Mejiab,c, Ting Zhoud, Aaron M. Ringd, Igor E. Brodskya, Christopher A. Huntera, and Boris Striepena,2 aDepartment of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104; bDepartment of Pathology and Laboratory Medicine, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; cDivision of Protective Immunity, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104; and dDepartment of Immunology, Yale School of Medicine, Yale University, New Haven, CT 06519 Edited by Stephen M. Beverley, Washington University School of Medicine, St. Louis, MO, and approved December 1, 2020 (received for review April 24, 2020) The apicomplexan parasite Cryptosporidium infects the intestinal Murine infection with C. tyzzeri resembles human cryptospo- epithelium. While infection is widespread around the world, chil- ridiosis in location, pathology, and resolution and provides an dren in resource-poor settings suffer a disproportionate disease important tool to define the host and parasite factors that burden. Cryptosporidiosis is a leading cause of diarrheal disease, determine the outcome of infection and to identify the im- responsible for mortality and stunted growth in children. CD4 mune
    [Show full text]
  • Post-Transcriptional Inhibition of Luciferase Reporter Assays
    THE JOURNAL OF BIOLOGICAL CHEMISTRY VOL. 287, NO. 34, pp. 28705–28716, August 17, 2012 © 2012 by The American Society for Biochemistry and Molecular Biology, Inc. Published in the U.S.A. Post-transcriptional Inhibition of Luciferase Reporter Assays by the Nod-like Receptor Proteins NLRX1 and NLRC3* Received for publication, December 12, 2011, and in revised form, June 18, 2012 Published, JBC Papers in Press, June 20, 2012, DOI 10.1074/jbc.M111.333146 Arthur Ling‡1,2, Fraser Soares‡1,2, David O. Croitoru‡1,3, Ivan Tattoli‡§, Leticia A. M. Carneiro‡4, Michele Boniotto¶, Szilvia Benko‡5, Dana J. Philpott§, and Stephen E. Girardin‡6 From the Departments of ‡Laboratory Medicine and Pathobiology and §Immunology, University of Toronto, Toronto M6G 2T6, Canada, and the ¶Modulation of Innate Immune Response, INSERM U1012, Paris South University School of Medicine, 63, rue Gabriel Peri, 94276 Le Kremlin-Bicêtre, France Background: A number of Nod-like receptors (NLRs) have been shown to inhibit signal transduction pathways using luciferase reporter assays (LRAs). Results: Overexpression of NLRX1 and NLRC3 results in nonspecific post-transcriptional inhibition of LRAs. Conclusion: LRAs are not a reliable technique to assess the inhibitory function of NLRs. Downloaded from Significance: The inhibitory role of NLRs on specific signal transduction pathways needs to be reevaluated. Luciferase reporter assays (LRAs) are widely used to assess the Nod-like receptors (NLRs)7 represent an important class of activity of specific signal transduction pathways. Although pow- intracellular pattern recognition molecules (PRMs), which are erful, rapid and convenient, this technique can also generate implicated in the detection and response to microbe- and dan- www.jbc.org artifactual results, as revealed for instance in the case of high ger-associated molecular patterns (MAMPs and DAMPs), throughput screens of inhibitory molecules.
    [Show full text]
  • NOD-Like Receptors in the Eye: Uncovering Its Role in Diabetic Retinopathy
    International Journal of Molecular Sciences Review NOD-like Receptors in the Eye: Uncovering Its Role in Diabetic Retinopathy Rayne R. Lim 1,2,3, Margaret E. Wieser 1, Rama R. Ganga 4, Veluchamy A. Barathi 5, Rajamani Lakshminarayanan 5 , Rajiv R. Mohan 1,2,3,6, Dean P. Hainsworth 6 and Shyam S. Chaurasia 1,2,3,* 1 Ocular Immunology and Angiogenesis Lab, University of Missouri, Columbia, MO 652011, USA; [email protected] (R.R.L.); [email protected] (M.E.W.); [email protected] (R.R.M.) 2 Department of Biomedical Sciences, University of Missouri, Columbia, MO 652011, USA 3 Ophthalmology, Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO 652011, USA 4 Surgery, University of Missouri, Columbia, MO 652011, USA; [email protected] 5 Singapore Eye Research Institute, Singapore 169856, Singapore; [email protected] (V.A.B.); [email protected] (R.L.) 6 Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO 652011, USA; [email protected] * Correspondence: [email protected]; Tel.: +1-573-882-3207 Received: 9 December 2019; Accepted: 27 January 2020; Published: 30 January 2020 Abstract: Diabetic retinopathy (DR) is an ocular complication of diabetes mellitus (DM). International Diabetic Federations (IDF) estimates up to 629 million people with DM by the year 2045 worldwide. Nearly 50% of DM patients will show evidence of diabetic-related eye problems. Therapeutic interventions for DR are limited and mostly involve surgical intervention at the late-stages of the disease. The lack of early-stage diagnostic tools and therapies, especially in DR, demands a better understanding of the biological processes involved in the etiology of disease progression.
    [Show full text]
  • Pattern Recognition Receptors in Health and Diseases
    Signal Transduction and Targeted Therapy www.nature.com/sigtrans REVIEW ARTICLE OPEN Pattern recognition receptors in health and diseases Danyang Li1,2 and Minghua Wu1,2 Pattern recognition receptors (PRRs) are a class of receptors that can directly recognize the specific molecular structures on the surface of pathogens, apoptotic host cells, and damaged senescent cells. PRRs bridge nonspecific immunity and specific immunity. Through the recognition and binding of ligands, PRRs can produce nonspecific anti-infection, antitumor, and other immunoprotective effects. Most PRRs in the innate immune system of vertebrates can be classified into the following five types based on protein domain homology: Toll-like receptors (TLRs), nucleotide oligomerization domain (NOD)-like receptors (NLRs), retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), C-type lectin receptors (CLRs), and absent in melanoma-2 (AIM2)-like receptors (ALRs). PRRs are basically composed of ligand recognition domains, intermediate domains, and effector domains. PRRs recognize and bind their respective ligands and recruit adaptor molecules with the same structure through their effector domains, initiating downstream signaling pathways to exert effects. In recent years, the increased researches on the recognition and binding of PRRs and their ligands have greatly promoted the understanding of different PRRs signaling pathways and provided ideas for the treatment of immune-related diseases and even tumors. This review describes in detail the history, the structural characteristics, ligand recognition mechanism, the signaling pathway, the related disease, new drugs in clinical trials and clinical therapy of different types of PRRs, and discusses the significance of the research on pattern recognition mechanism for the treatment of PRR-related diseases.
    [Show full text]
  • Supplementary Table 2 Supplementary Table 1
    Supplementary table 1 Rai/ Binet IGHV Cytogenetic Relative viability Fludarabine- Sex Outcome CD38 (%) IGHV gene ZAP70 (%) Treatment (s) Stage identity (%) abnormalities* increase refractory 1 M 0/A Progressive 14,90 IGHV3-64*05 99,65 28,20 Del17p 18.0% 62,58322819 FCR n.a. 2 F 0/A Progressive 78,77 IGHV3-48*03 100,00 51,90 Del17p 24.8% 77,88052021 FCR n.a. 3 M 0/A Progressive 29,81 IGHV4-b*01 100,00 9,10 Del17p 12.0% 36,48 Len, Chl n.a. 4 M 1/A Stable 97,04 IGHV3-21*01 97,22 18,11 Normal 85,4191657 n.a. n.a. Chl+O, PCR, 5 F 0/A Progressive 87,00 IGHV4-39*07 100,00 43,20 Del13q 68.3% 35,23314039 n.a. HDMP+R 6 M 0/A Progressive 1,81 IGHV3-43*01 100,00 20,90 Del13q 77.7% 57,52490626 Chl n.a. Chl, FR, R-CHOP, 7 M 0/A Progressive 97,80 IGHV1-3*01 100,00 9,80 Del17p 88.5% 48,57389901 n.a. HDMP+R 8 F 2/B Progressive 69,07 IGHV5-a*03 100,00 16,50 Del17p 77.2% 107,9656878 FCR, BA No R-CHOP, FCR, 9 M 1/A Progressive 2,13 IGHV3-23*01 97,22 29,80 Del11q 16.3% 134,5866919 Yes Flavopiridol, BA 10 M 2/A Progressive 0,36 IGHV3-30*02 92,01 0,38 Del13q 81.9% 78,91844953 Unknown n.a. 11 M 2/B Progressive 15,17 IGHV3-20*01 100,00 13,20 Del11q 95.3% 75,52880995 FCR, R-CHOP, BR No 12 M 0/A Stable 0,14 IGHV3-30*02 90,62 7,40 Del13q 13.0% 13,0939004 n.a.
    [Show full text]
  • ATP-Binding and Hydrolysis in Inflammasome Activation
    molecules Review ATP-Binding and Hydrolysis in Inflammasome Activation Christina F. Sandall, Bjoern K. Ziehr and Justin A. MacDonald * Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada; [email protected] (C.F.S.); [email protected] (B.K.Z.) * Correspondence: [email protected]; Tel.: +1-403-210-8433 Academic Editor: Massimo Bertinaria Received: 15 September 2020; Accepted: 3 October 2020; Published: 7 October 2020 Abstract: The prototypical model for NOD-like receptor (NLR) inflammasome assembly includes nucleotide-dependent activation of the NLR downstream of pathogen- or danger-associated molecular pattern (PAMP or DAMP) recognition, followed by nucleation of hetero-oligomeric platforms that lie upstream of inflammatory responses associated with innate immunity. As members of the STAND ATPases, the NLRs are generally thought to share a similar model of ATP-dependent activation and effect. However, recent observations have challenged this paradigm to reveal novel and complex biochemical processes to discern NLRs from other STAND proteins. In this review, we highlight past findings that identify the regulatory importance of conserved ATP-binding and hydrolysis motifs within the nucleotide-binding NACHT domain of NLRs and explore recent breakthroughs that generate connections between NLR protein structure and function. Indeed, newly deposited NLR structures for NLRC4 and NLRP3 have provided unique perspectives on the ATP-dependency of inflammasome activation. Novel molecular dynamic simulations of NLRP3 examined the active site of ADP- and ATP-bound models. The findings support distinctions in nucleotide-binding domain topology with occupancy of ATP or ADP that are in turn disseminated on to the global protein structure.
    [Show full text]
  • NLRP1 Haplotypes Associated with Vitiligo and Autoimmunity Increase Interleukin-1Β Processing Via the NLRP1 Inflammasome
    NLRP1 haplotypes associated with vitiligo and autoimmunity increase interleukin-1β processing via the NLRP1 inflammasome Cecilia B. Levandowskia, Christina M. Maillouxa, Tracey M. Ferraraa, Katherine Gowana, Songtao Bena, Ying Jina,b, Kimberly K. McFannc, Paulene J. Hollanda, Pamela R. Faina,b,d, Charles A. Dinarelloe,1, and Richard A. Spritza,b,1 aHuman Medical Genetics and Genomics Program and bDepartment of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045; cColorado Biostatistics Consortium, Colorado School of Public Health, Aurora, CO 80045; and dBarbara Davis Center for Childhood Diabetes, and eDepartment of Medicine, University of Colorado School of Medicine, Aurora, CO 80045 Contributed by Charles A. Dinarello, January 8, 2013 (sent for review December 26, 2012) Nuclear localization leucine-rich-repeat protein 1 (NLRP1) is a key associated molecular patterns stimulate surface Toll-like recep- regulator of the innate immune system, particularly in the skin tors (TLRs) with subsequent assembly of the NLRP1 inflam- where, in response to molecular triggers such as pathogen- masome and activation of caspase-1. Active caspase-1 then associated or damage-associated molecular patterns, the NLRP1 cleaves the inactive IL-1β precursor (pro–IL-1β) to the mature inflammasome promotes caspase-1–dependent processing of bio- bioactive IL-1β (23), thereby stimulating downstream inflam- active interleukin-1β (IL-1β), resulting in IL-1β secretion and down- matory responses (23, 24). Because of its highly inflammatory stream inflammatory responses. NLRP1 is genetically associated and immunostimulating properties, the processing and release of with risk of several autoimmune diseases including generalized bioactive IL-1β are tightly controlled. However, in cryopyrin- vitiligo, Addison disease, type 1 diabetes, rheumatoid arthritis, associated periodic syndromes (CAPSs), which include familial and others.
    [Show full text]
  • Coincidental Loss of DOCK8 Function in NLRP10-Deficient and C3H/Hej Mice Results in Defective Dendritic Cell Migration
    Coincidental loss of DOCK8 function in NLRP10-deficient and C3H/HeJ mice results in defective dendritic cell migration Jayendra Kumar Krishnaswamya,b,1, Arpita Singha,b,1, Uthaman Gowthamana,b, Renee Wua,b, Pavane Gorrepatia,b, Manuela Sales Nascimentoa,b, Antonia Gallmana,b, Dong Liua,b, Anne Marie Rhebergenb, Samuele Calabroa,b, Lan Xua,b, Patricia Ranneya,b, Anuj Srivastavac, Matthew Ransond, James D. Gorhamd, Zachary McCawe, Steven R. Kleebergere, Leonhard X. Heinzf, André C. Müllerf, Keiryn L. Bennettf, Giulio Superti-Furgaf, Jorge Henao-Mejiag, Fayyaz S. Sutterwalah, Adam Williamsi, Richard A. Flavellb,j,2, and Stephanie C. Eisenbartha,b,2 Departments of aLaboratory Medicine and bImmunobiology and jHoward Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520; cComputational Sciences, The Jackson Laboratory, Bar Harbor, ME 04609; dDepartment of Pathology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755; eNational Institute of Environmental Health Sciences, Research Triangle Park, NC 27709; fCeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; gInstitute for Immunology, Departments of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; hInflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA 52241; and iThe Jackson Laboratory for Genomic Medicine, Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032 Contributed by Richard A. Flavell, January 28, 2015 (sent for review December 23, 2014; reviewed by Matthew L. Albert and Thirumala-Devi Kanneganti) Dendritic cells (DCs) are the primary leukocytes responsible for NLRP10 is the only NOD-like receptor (NLR) without a leu- priming T cells.
    [Show full text]